Your browser doesn't support javascript.
loading
In vitro characterization of rare anti-αIIbß3 isoantibodies produced by patients with Glanzmann thrombasthenia that severely block fibrinogen binding and generate procoagulant platelets via complement activation.
Lee, Christine S M; Huguenin, Yoann; Pillois, Xavier; Moulieras, Mikeldi; Marcy, Ella; Whittaker, Shane; Chen, Vivien M Y; Fiore, Mathieu.
Afiliación
  • Lee CSM; ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia.
  • Huguenin Y; Competence Centre for Inherited Bleeding Disorders, University Hospital of Bordeaux, Bordeaux, France.
  • Pillois X; French Reference Centre for Inherited Platelet Disorders, University Hospital of Bordeaux, Pessac, France.
  • Moulieras M; French Reference Centre for Inherited Platelet Disorders, University Hospital of Bordeaux, Pessac, France.
  • Marcy E; French Reference Centre for Inherited Platelet Disorders, University Hospital of Bordeaux, Pessac, France.
  • Whittaker S; ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia.
  • Chen VMY; ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia.
  • Fiore M; Department of Haematology, Concord Repatriation General Hospital and NSW Health Pathology, Sydney, New South Wales, Australia.
Res Pract Thromb Haemost ; 8(1): 102253, 2024 Jan.
Article en En | MEDLINE | ID: mdl-38268518
ABSTRACT

Background:

Glanzmann thrombasthenia (GT) is a rare bleeding disorder caused by inherited defects of the platelet αIIbß3 integrin. Platelet transfusions can be followed by an immune response that can block integrin function by interfering with fibrinogen binding.

Objectives:

In this study, we aimed to determine the prevalence of such isoantibodies and better characterize their pathogenic properties.

Methods:

Twelve patients with GT were evaluated for anti-αIIbß3 isoantibodies. Sera from patients with GT with or without anti-αIIbß3 isoantibodies were then used to study their in vitro effect on platelets from healthy donors. We used several approaches (IgG purification, immunofluorescence staining, and inhibition of signaling pathways) to characterize the pathogenic properties of the anti-αIIbß3 isoantibodies.

Results:

Only 2 samples were able to severely block integrin function. We observed that these 2 sera caused a reduction in platelet size similar to that observed when platelets become procoagulant. Mixing healthy donor platelets with patients' sera or purified IgGs led to microvesiculation, phosphatidylserine exposure, and induction of calcium influx. This was associated with an increase in procoagulant platelets. Pore formation and calcium entry were associated with complement activation, leading to the constitution of a membrane attack complex (MAC) with enhanced complement protein C5b-9 formation. This process was inhibited by the complement 5 inhibitor eculizumab and reduced by polyvalent human immunoglobulins.

Conclusion:

Our data suggest that complement activation induced by rare blocking anti-αIIbß3 isoantibodies may lead to the formation of a MAC with subsequent pore formation, resulting in calcium influx and procoagulant platelet phenotype.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Risk_factors_studies Idioma: En Revista: Res Pract Thromb Haemost Año: 2024 Tipo del documento: Article País de afiliación: Australia Pais de publicación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Risk_factors_studies Idioma: En Revista: Res Pract Thromb Haemost Año: 2024 Tipo del documento: Article País de afiliación: Australia Pais de publicación: Estados Unidos